Anda di halaman 1dari 9

Myocardial Osteopontin Expression Coincides With the

Development of Heart Failure


Krishna Singh, Geza Sirokman, Catherine Communal, Kathleen G. Robinson, Chester H. Conrad,
Wesley W. Brooks, Oscar H.L. Bing, Wilson S. Colucci
AbstractTo identify genes that are differentially expressed during the transition from compensated hypertrophy to failure,
myocardial mRNA from spontaneously hypertensive rats (SHR) with heart failure (SHR-F) was compared with that
from age-matched SHR with compensated hypertrophy (SHR-NF) and normotensive Wistar-Kyoto rats (WKY) by
differential display reverse transcriptasepolymerase chain reaction. Characterization of a transcript differentially
expressed in SHR-F yielded a cDNA with homology to the extracellular matrix protein osteopontin. Northern analysis
showed low levels of osteopontin mRNA in left ventricular myocardium from WKY and SHR-NF but a markedly
increased ('10-fold) level in SHR-F. In myocardium from WKY and SHR-NF, in situ hybridization showed only scant
osteopontin mRNA, primarily in arteriolar cells. In SHR-F, in situ hybridization revealed abundant expression of
osteopontin mRNA, primarily in nonmyocytes in the interstitial and perivascular space. Similar findings for osteopontin
protein were observed in the midwall region of myocardium from the SHR-F group. Consistent with the findings in
SHR, osteopontin mRNA was minimally increased ('1.9-fold) in left ventricular myocardium from nonfailing
aortic-banded rats with pressure-overload hypertrophy but was markedly increased ('8-fold) in banded rats with failure.
Treatment with captopril starting before or after the onset of failure in the SHR reduced the increase in left ventricular
osteopontin mRNA levels. Thus, osteopontin expression is markedly increased in the heart coincident with the
development of heart failure. The source of osteopontin in SHR-F is primarily nonmyocytes, and its induction is
inhibited by an angiotensin-converting enzyme inhibitor, suggesting a role for angiotensin II. Given the known
biological activities of osteopontin, including cell adhesion and regulation of inducible nitric oxide synthase gene
expression, these data suggest that it could play a role in the pathophysiology of heart failure. (Hypertension.
1999;33:663-670.)
Key Words: osteopontin n heart n heart failure n rats, inbred SHR n hypertrophy

he spontaneously hypertensive rat (SHR) has been


proven to be a valuable model for studying the transition
from myocardial hypertrophy to failure.1 4 In these animals,
a period of several months of compensated hypertrophy is
followed by the development of heart failure (SHR-F). We
have shown that in comparison to SHR with compensated
hypertrophy (SHR-NF), the transition is associated with
pathological evidence of cardiac decompensation, ie, presence of pleuropericardial effusions, atrial thrombi, and right
ventricular (RV) hypertrophy with reduced contractile function and increased stiffness of papillary muscle, increased
interstitial fibrosis, and increased expression of atrial natriuretic peptide (ANP), collagen, fibronectin, and transforming
growth factor-b1 mRNAs.3,5 8
In an attempt to identify genes that are differentially
expressed coincident with the transition from hypertrophy to
failure, we used differential display reverse transcriptase
polymerase chain reaction (RT-PCR) to compare RNA iso-

lated from the left ventricles (LV) of SHR-F with that from
age-matched SHR-NF and Wistar-Kyoto rats (WKY).9 One
of the transcripts that was differentially expressed in myocardium from SHR-F was for osteopontin, an extracellular
matrix protein that can act as an adhesion molecule, affect
cellular function by interacting with integrins, and modulate
the expression of inducible nitric oxide synthase.10 12
Although it has recently been shown that osteopontin can
be expressed in myocardium in response to necrotic injury13
or short-term pressure overload,14,15 its expression has not
previously been associated with the development of myocardial failure. This report describes the differential expression
and localization of osteopontin in the myocardium of both
SHR and aortic-banded rats coincident with the development
of pathological evidence of cardiac decompensation. Since
angiotensin II (Ang II) stimulates osteopontin expression by
cardiac fibroblasts14 and angiotensin-converting enzyme
(ACE) inhibitor can prevent many of the features of myocar-

Received June 24, 1998; first decision July 14, 1998; revision accepted October 16, 1998.
From the Myocardial Biology Unit and Cardiovascular Division, Department of Medicine, Boston Medical Center, Boston Veterans Affairs Medical
Center and Boston University School of Medicine, Boston, Mass.
Correspondence to Krishna Singh, PhD, Myocardial Biology Unit, Boston University School of Medicine, 80 E Concord St, Boston, MA 02118. E-mail
krishna.singh@bmc.org
1999 American Heart Association, Inc.
Hypertension is available at http://www.hypertensionaha.org

663
Downloaded from http://hyper.ahajournals.org/
by guest on August 24, 2015

664

Myocardial Osteopontin
Body and Ventricular Weights
WKY (n56)

Nonfailing SHR (n55)

Failing SHR (n58)

SHR
Body weight, g

566623

378632*

383618*

LV weight, g

1.2760.1

1.3360.12

1.3960.06

RV weight, g

0.360.03

0.2460.02

0.4660.02*

LV/body weight, mg/g

2.2460.09

3.5160.17*

3.6360.04*

RV/body weight, mg/g

0.5360.04

0.6360.02

1.2060.08*

WKY (n55)

Nonfailing Banded (n55)

Failing Banded (n55)

Aorticbanded rats
Body weight, g

573647

603631

594623

LV weight, g

1.0360.05

1.5460.04*

1.760.07*

RV weight, g

0.2760.01

0.3160.01

0.5660.02*

LV/body weight, mg/g

1.8060.07

2.5560.13*

2.8660.02*

RV/body weight, mg/g

0.4760.02

0.5160.03

0.9460.05*

*P,0.01 vs WKY.
P,0.01 vs nonfailing SHR or banded rat.

dial failure in SHR,1,2 we also examined the effect of


treatment with the ACE inhibitor captopril on the expression
of osteopontin.

Methods

the RNA was extracted with phenol-chloroform, the RNA was


precipitated with ethanol at 220C. For differential display RTPCR, RNA was treated with RNase-free DNase for 30 minutes at
37C. The RNA was extracted again with phenol-chloroform and
precipitated with ethanol.

Differential Display and Sequence Analysis

Animal Models
Male SHR and nonhypertensive control WKY were purchased
(Taconic, Germantown, NY) at the age of 6 to 9 months and boarded
until the time of study. Beginning at 18 months of age, all animals
were observed twice per week for tachypnea and labored respiration.2,5,7,8 SHR were studied within 7 days of the observation of
respiratory difficulties. Age-matched WKY and SHR without these
signs were studied at the same time.
The aortic-banded rats and sham-operated WKY were purchased
from Taconic, Germantown, NY (surgeries were performed at
Taconic). A loop of 3.0 surgical silk was placed around the aortic
arch between the brachiocephalic and left carotid artery. A blunted
18.5-gauge needle was placed over the aorta, and the silk was tied
around the needle and aorta.16 The needle was then carefully
removed, resulting in a nonconstricting band. The development of
heart failure was suggested by the observation of respiratory difficulties and documented by pathological examination, as described
for the SHR.2,5,7,8 The failing and nonfailing banded animals and
their age-matched nonbanded controls were studied at 11 months of
age, '9 months after banding.
Captopril was added to the drinking water (2.0 g/L) of 9 SHR, as
previously described.2,17 In 4 animals, it was added before the onset
of failure at either 12 (n51), 18 (n51), or 21 (n52) months of age
and continued until the age of 24 months. In 5 rats, captopril was
administered after the onset of failure and continued for 2 to 4
months before euthanasia at the age of 24 months.
After the rats were killed, the hearts were quickly removed and
placed in Krebs-Henseleit buffer. The RV and LV were carefully
dissected, weighed, and immediately frozen in liquid nitrogen for
RNA isolation. For in situ hybridization and immunohistochemical
studies, hearts were perfusion-fixed with freshly prepared 4%
paraformaldehyde.6

Differential display and sequence analysis were performed as described earlier. 9 With the use of anchored primers 59AAGCTTTTTTTTTTTG-39, 59-AAGCTTTTTTTTTTTC-39, or 59AAGCTTTTTTTTTTTA-39 and Moloney murine leukemia virus
reverse transcriptase (GeneHunter Co), the cDNAs were prepared

Figure 1. Representative autoradiograph showing differential display RT-PCR of RNA samples isolated from the LV of age-matched
WKY and SHR-F or SHR-NF. The
oligonucleotides consisted of an
anchor primer
59-AAGCTTTTTTTTTTTC-39 and an
upstream primer
59-AAGCTTCGACTGT. The arrow
indicates a band that is increased
in SHR-F compared with SHR-NF
or WKY. The band from the SHR-F
was excised from the gel, cloned,
and sequenced.

RNA Isolation
Total RNA was isolated from the LV according to the method of
Chomczynski and Sacchi.18 Briefly, samples (100 to 200 mg) were
homogenized in guanidinium thiocyanate solution (4 mol/L guanidinium thiocyanate, 25 mmol/L Na citrate [pH 7.0], 0.5% sarcosyl,
and 0.1 mol/L 2-mercaptoethanol) with a tissue homogenizer. After

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

Singh et al

February 1999

665

Figure 2. Alignment of osteopontin cDNA


sequence from rat (accession No. M99252)19 and
the 129-bp fragment sequenced from the 500-bp
cDNA identified by differential display RT-PCR of
LV myocardial mRNA from SHR-F.

from mRNA of total RNA isolated from the LV. The reverse
transcription mixture was then amplified by random priming PCR
containing [a-35S]dATP with the appropriate anchored primer and 24
different arbitrary 13-mer primers (GeneHunter Co). The products
were separated on a polyacrylamide sequencing gel containing 6
mol/L urea. Autoradiographs were evaluated by visual comparison
of the intensities of individual bands run side by side.
The differentially expressed cDNAs were excised from the dried
gel and extracted with hot water. The recovered DNA was reamplified and cloned in a pCR 2.1 vector (Invitrogen). Purified plasmid
DNA from Escherichia coli was then used for sequencing according
to the dideoxy chain termination method with Sequanase 2.0 (Amersham). Sequences were compared with a sequence database with
the use of the Geneworks program (Intelligenetics).

Northern Blot Analysis


Total RNA was size fractionated on 1.0% formaldehyde agarose gels
containing 2.2 mol/L formaldehyde and transferred to nylon membranes (Gene Screen Plus; NEN DuPont) with a transblot apparatus
(Bio-Rad). Blots were hybridized overnight with the radiolabeled
osteopontin cDNA probe,19 as described previously.10 The blots
were then deprobed and hybridized with radiolabeled ANP cDNA
(courtesy of Dr C. Seidman) probe. To normalize for loading
differences, the blots were then probed with an 18S oligonucleotide
(30-mer) end-labeled by T4 polynucleotide kinase.10 Differences in
mRNA signal intensity were determined with the use of a phosphoimager (PhosphoImager, Bio-Rad).

In Situ Hybridization
In situ hybridization was performed as previously described.6 Hearts
were perfusion fixed with 4% paraformaldehyde/phosphate-buffered
saline. Parallel slices 1 to 2 mm thick, encompassing both RV and
LV, were dehydrated and embedded in Paraplast Plus embedding
medium (Oxford). Sections 4 mm thick (from WKY, SHR-NF, and
SHR-F) were hybridized with single-stranded sense or antisense
RNA probes transcribed from a linearized20 full-length osteopontin
cDNA19 using [a-35S]UTP. Probes were extracted with phenolchloroform and precipitated with ethanol.

Immunohistochemistry
Sections (4 mm thick) from WKY, SHR-NF, and SHR-F were
deparaffinized and stained with the use of monoclonal antiosteopontin antibodies (MPIIIB10; developmental studies, hybridoma bank) and Vectastain avidin-biotin peroxidase kit (Vector
Laboratories). Briefly, nonspecific binding was minimized by incubation for 20 minutes with 1.5% normal horse serum. The sections
were then incubated for 30 minutes with MPIIIB10 (1:250). After
they were washed, the sections were incubated with biotinylated
secondary antibodies. Detection was performed with the use of
Vectastain ABC-AP reagent and Vector Red alkaline substrate kit
(Vector Laboratories). The sections were visualized and photographed under epifluorescence microscopy with the use of rhodamine excitation and emission filters.

Statistical Analysis
All data are expressed as mean6SEM. Two-tailed Students t test
was used to compare the group means. Probability values of ,0.05
were considered significant.

Results
Figure 3. Increased expression of osteopontin mRNA in the LV
of SHR-F. A, Representative autoradiograph of a Northern
hybridization with a cDNA for osteopontin in LV myocardium
obtained from WKY, SHR-NF, and SHR-F. ANP mRNA was
moderately increased in SHR-NF and further increased in
SHR-F. B, Mean data for osteopontin mRNA in LV from WKY
(n56), SHR-NF (n55), SHR-F (n58), and SHR in which captopril
treatment was begun before (CPT-1) ( n54) or after (CPT-2)
(n55) the development of clinical failure. *P,0.05 vs WKY;
P,0.05 vs SHR-NF; #P,0.05 vs SHR-F; P50.067 vs SHR-F.

Pathophysiologic Data
The body, LV, and RV weights for each experimental group
are shown in the Table. SHR is well documented to develop
cardiac decompensation between 18 and 24 months.2 4,6,9 The
LV/body weight ratio was increased to a similar degree
('50% to 60%) in the SHR-F and SHR-NF groups. The
RV/body weight ratio was higher in SHR-F than in SHR-NF,
consistent with prior findings in these animals. Among the

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

666

Myocardial Osteopontin

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

Singh et al

February 1999

667

Figure 6. Expression of osteopontin mRNA in LV of rats with


failure caused by chronic aortic banding. A, Representative
autoradiograph of Northern hybridization with a cDNA for
osteopontin in LV myocardium obtained from control WKY without aortic banding (WKY), banded rats without signs of failure
(AOB-NF), and banded rats with signs of failure (AOB-F).
Osteopontin mRNA was slightly increased in nonfailing banded
rats and markedly increased in failing rats. ANP mRNA was
moderately increased in nonfailing banded rats and further
increased in failing rats. B, Mean data from 5 WKY, 5 AOB-NF,
and 5 AOB-F. *P50.1 vs WKY; #P,0.05 vs WKY; P,0.05 vs
AOB-NF.

Differential Display RT-PCR

Figure 5. Immunohistochemical staining of LV myocardium


using MPIIIB10. A shows the midwall area from SHR-NF, and B
and C show the midwall area from SHR-F at magnifications of
3300 and 3600, respectively. The orange staining for
osteopontin (arrows) was observed mainly in the interstitial
space.

SHR-F, all animals also showed pleuropericardial effusions


and atrial thrombi, whereas these findings were absent among
the SHR-NF and WKY groups.
In aortic-banded animals, the LV/body weight ratio was
increased to a similar degree ('40% to 60%) in both the
nonfailing and failing banded animals (versus nonbanded
control animals), whereas the RV/body weight ratio was
increased only in failing banded animals, similar to the
findings in SHR.

To identify genes with differential expression in LV myocardium from SHR-F (versus SHR-NF and age-matched WKY),
3 hearts from each group were subjected to differential
display RT-PCR. An autoradiograph of a differential display
gel showed the increased expression of a cDNA in SHR-F
versus SHR-NF and WKY (Figure 1). This cDNA ('500 bp)
was isolated and cloned, and 129 bp were sequenced, revealing .98% homology to the rat osteopontin cDNA sequence
(Figure 2).19

Northern Hybridization
The differential expression of osteopontin mRNA in SHR-F
was confirmed by performing Northern hybridization with
total RNA isolated from the LV of age-matched SHR-F,
SHR-NF, and WKY. Hybridization with a 1.5-kb osteopontin
cDNA identified a 1.6-kb transcript similar in size to that
reported for osteopontin.10 Osteopontin mRNA was expressed at similar low levels in WKY and SHR-NF (Figure
3). In striking contrast, osteopontin mRNA was increased
10.262.57-fold (P,0.05; n58) in LV from SHR-F versus

Figure 4. In situ hybridization with an antisense osteopontin cRNA probe. Sections from SHR-NF (A and C) and SHR-F (B and D) were
hybridized with an antisense probe and photographed under darkfield illumination (magnification 3100). A and B show the endocardial
area, and C and D show the midwall area. E through H are sections from SHR-F hybridized with the antisense probe, stained with
hematoxylin and eosin, and photographed (magnification 3400).

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

668

Myocardial Osteopontin

SHR-NF (Figure 3). One of 8 hearts in the SHR-F group had


osteopontin mRNA values that were similar to those in the
SHR-NF group. ANP mRNA was increased '10-fold in
SHR-NF versus WKY. In SHR-F, ANP mRNA was further
increased to .70-fold versus WKY and '7-fold versus
SHR-NF (Figure 3).

In Situ Hybridization
In sections from SHR-NF hearts, in situ hybridization with
the antisense probe for osteopontin showed scant expression,
which was limited to blood vessels, possibly in endothelial
and/or smooth muscle cells (Figure 4A and 4C). Similarly,
WKY hearts showed scant expression of osteopontin mRNA
(not shown). In striking contrast, sections from SHR-F hearts
revealed intense expression of osteopontin, primarily in the
interstitial and perivascular space (Figure 4B and 4D). Hematoxylin and eosin staining of adjacent sections confirmed
that osteopontin expression in SHR-F was associated primarily with nonmuscle cells infiltrating between the myocytes
(Figure 4E through 4H). In some areas, the expression of
osteopontin was diffuse within the interstitial space (eg,
Figure 4F and 4H). In other areas, the expression was more
focal and appeared to be associated with degenerating myocytes (eg, Figure 4E and 4G). Increased expression of
osteopontin mRNA was also detected in the RV of SHR-F, in
a distribution similar to that observed in the LV. No grains
were visible with the sense osteopontin probe (not shown).

Immunohistochemistry
In sections from SHR-NF hearts, immunohistochemical analysis demonstrated low levels of immunoreactivity for osteopontin in the interstitial cells of the midwall region of LV
(Figure 5A). Similar staining for osteopontin was observed in
sections from WKY (not shown). In sections from SHR-F,
intense staining was observed in the midwall region of LV,
primarily in the interstitial space (Figure 5B). Most of the
staining was observed in the areas of fibrosis and seemed to
be present around the myocytes (Figure 5C). The papillary
muscles from the hearts of WKY, SHR-NF, and SHR-F all
showed intense fibrosis associated with marked staining for
osteopontin.

Effect of Captopril on Osteopontin Expression


in SHR
The effect of treatment with the ACE inhibitor captopril on
LV osteopontin mRNA expression was assessed by Northern
hybridization in 9 SHR at 24 months of age. Captopril
prevented the increase in osteopontin expression in 4 of 4
hearts treated before failure, while it reduced osteopontin
expression in 4 of 5 hearts from animals treated after the onset
of failure (Figure 3B).

Osteopontin Expression in Aortic-Banded Rats


Compared with age-matched WKY controls, osteopontin
mRNA was increased 1.960.6-fold (P50.1 versus WKY;
n55) in banded animals without failure and 7.762.9-fold
(P,0.05 versus nonfailing banded; n55) in banded animals
with signs of failure (Figure 6). ANP was increased 3.460.9-

fold and 5.261.1-fold in nonfailing and failing banded


animals, respectively.

Discussion
Using differential display RT-PCR and Northern analysis, we
found that the expression of osteopontin mRNA was markedly increased coincident with appearance of pathophysiological evidence of cardiac decompensation in 2 models of
chronic pressure overload, the SHR and the aortic-banded rat.
These results, confirmed by in situ hybridization and immunohistochemistry, indicated that (1) there was only low basal
expression of osteopontin in control WKY rats; (2) in the
absence of failure, basal expression of osteopontin was
unchanged or only slightly increased in SHR or aortic-banded
rats with well-established LV hypertrophy; and (3) in both
models there was a marked increase in osteopontin expression in age-matched animals that had manifestations of
cardiac decompensation. In situ hybridization and immunohistochemistry demonstrated that in failing animals the primary source of osteopontin mRNA was nonmyocytes and
that the protein is mainly localized in the interstitium.
Osteopontin is an arginine-glycine-aspartate containing
adhesive glycoprotein. Although first isolated from mineralized bone matrix, osteopontin can be synthesized by several
cell types, including cardiac myocytes, microvascular endothelial cells, and fibroblasts.10 12,14,21,22 Osteopontin is expressed in several tissues in response to injury, suggesting a
role in the reparative process, and appears capable of mediating diverse biological functions, including cell adhesion,
chemotaxis, and signaling.12,13,23,24
Several groups have recently demonstrated that osteopontin can be expressed in the myocardium. Murry et al13 showed
the expression of osteopontin by macrophages in response to
myocardial necrosis caused by transdiaphragmatic freezing.
Likewise, in the cardiomyopathic Syrian hamster, Williams et
al23 found expression of osteopontin in areas of necrosis
associated with inflammation, also apparently in tissue macrophages. Recently, Graf et al15 demonstrated that osteopontin mRNA was increased approximately 2-fold and 3-fold,
respectively, in LV from rats with 2-kidney, 1 clip hypertension or aortic banding. In these 2 models, cardiac myocytes
were identified as the predominant source of osteopontin by
immunohistochemistry and in situ hybridization.15
In contrast to Graf et al,15 we did not find osteopontin
mRNA to be increased in the LV of SHR-NF as assessed by
differential display PCR, Northern hybridization, or in situ
hybridization, despite clear evidence of LV hypertrophy and
the expression of ANP mRNA. Likewise, we found only a
modest ('1.9-fold) increase in osteopontin in nonfailing
aortic-banded rats, despite marked LV hypertrophy and
increased ANP mRNA expression. A possible explanation for
differences in findings between our studies and those of Graf
et al15 may relate to the markedly different time periods
studied. Whereas Graf et al15 examined animals relatively
soon ('4 to 6 weeks) after surgery, our studies were
performed in animals that had been exposed to LV pressure
overload for many months (18 to 24 months for SHR; .9
months for aortic-banded rats).

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

Singh et al
Consistent with the in situ hybridization, immunohistochemistry revealed increased expression of osteopontin in the
midwall region of SHR-F (compared with SHR-NF and
WKY). Similar to the in situ hybridization, most of the
staining was observed in the interstitial space. In contrast to in
situ hybridization, which showed increased expression of
osteopontin mRNA in the papillary muscles of only SHR-F,
immunoreactivity for osteopontin protein was increased in
the papillary muscles of all 3 groups (ie, WKY, SHR-NF, and
SHR-F). This may reflect (1) the extensive fibrosis in
age-related papillary muscles from all 3 groups and/or (2) that
secretory osteopontin, once incorporated into extracellular
matrix, has a lower turnover rate and/or higher stability.
In SHR with cardiac decompensation, the major source of
osteopontin expression appears to be nonmyocytes in the
interstitium and perivascular space. Thus, our data differ from
those of Graf et al,15 who found that myocytes were the major
source of osteopontin in LV from animals with relatively
short-term hypertrophy. However, our findings are consistent
with those of Murry et al13 and Williams et al,23 who found
interstitial expression of osteopontin by macrophages in rats
with thermal injury or cardiomyopathic hamsters, respectively. Murry et al13 suggested that osteopontin is synthesized
in response to injury. Our findings suggest that, at least with
regard to osteopontin, the appearance of cardiac decompensation and the transition to failure in these rat models is
associated with an injury response. Taken together with the
data of Graf et al,15 it appears reasonable to suggest that the
source of osteopontin mRNA may be myocytes early in LV
hypertrophy but shifts to interstitial cells and focal areas of
myocyte injury in late hypertrophy with the development of
heart failure.
Ang II has been shown to cause focal myocyte necrosis in
rats infused with Ang II or in models of renovascular
hypertension.25 Ang II also induces osteopontin in cardiac
fibroblasts in vitro,14 and infusion of Ang II increased
osteopontin expression in kidney.26 In our study, focal expression of osteopontin mRNA appeared to be associated
with degenerating myocytes (Figure 4E and 4G). We also
found that captopril treatment of SHR reduced osteopontin
mRNA levels. These findings are thus consistent with a role
for Ang II in the induction of osteopontin in areas of myocyte
necrosis and fibrosis during the transition to failure. However, it is also possible that rats treated with captopril before
the development of heart failure remained compensated and
therefore showed no increase in osteopontin, while captopril
treatment after the development of heart failure might have
prevented the additional necrosis and healing of lesions,
thereby preventing osteopontin expression. Another possibility is that captopril suppressed the expression of osteopontin
by increasing bradykinin, which might act through nitric
oxide to attenuate the expression of growth-related genes.27
The role of osteopontin during the transition to heart failure
is not known. Indeed, relatively little is known about the role
of osteopontin in the myocardium. Osteopontin can act as an
adhesion molecule, as a chemotactic factor, and as a substrate
for the migration of macrophages, smooth muscle cells, and
endothelial cells.12 Osteopontin can also act as a cytokine to
stimulate lymphocyte immunoglobulin production.11,28 These

February 1999

669

activities are consistent with a role for osteopontin in fibrosis


or healing in response to injury.
We have also shown that osteopontin can suppress the
cytokine-induced expression of inducible nitric oxide synthase in cardiac myocytes and microvascular endothelial cells
isolated from adult rat hearts.10 It is therefore of interest that
high levels of nitric oxide produced by inducible nitric oxide
synthase can impair myocyte function29 and may be toxic to
cardiac myocytes.30 Since it appears that myocardial inducible nitric oxide synthase expression is increased with failure,31,32 osteopontin has the potential to modulate the effects
of inflammatory cytokines on the myocardium by attenuating
the expression of inducible nitric oxide synthase.
Thus, osteopontin expression is markedly increased in the
myocardium coincident with the development of evidence of
cardiac decompensation in 2 models of chronic pressure
overload. Given the importance of the extracellular matrix in
the pathophysiology of myocardial failure33,34 and the known
biological activities of osteopontin, it is possible that this
interstitial matrix protein plays an important role in the
pathogenesis of heart failure.

Acknowledgments
This study was supported in part by grants R29 HL-57947 (Dr Singh)
and HL-52320 and HL-42539 (Dr Colucci) from the National
Institutes of Health and by Medical Research Funds from the
Department of Veterans Affairs (Drs Singh, Bing, Conrad, and
Brooks). Dr Communal is supported by a fellowship from the
American Heart Association, Massachusetts Affiliate.

References
1. Lorell B. Transition from hypertrophy to failure. Circulation. 1997;96:
3824 3827.
2. Brooks WW, Bing OHL, Robinson KG, Slawsky MT, Chaletsky DM,
Conrad CH. Effects of angiotensin-converting enzyme inhibition on myocardial fibrosis and function in hypertrophied and failing myocardium
from the spontaneously hypertensive rat. Circulation. 1997;96:
4002 4010.
3. Boluyt MO, Bing OHL, Lakatta EG. The aging spontaneously hypertensive rat as a model of the transition from stable compensated hypertrophy to heart failure. Eur Heart J. 1996;16(suppl N):19 30.
4. Mitchell GF, Pfeffer JM, Pfeffer MA. The transition to failure in the
spontaneously hypertensive rat. Am J Physiol. 1997;10:120S126S.
5. Bing OHL, Brooks WW, Robinson KG, Slawsky MT, Hayes JA, Litwin
SE, Sen S, Conrad CH. The spontaneously hypertensive rat as a model of
the transition from compensated left ventricular hypertrophy to failure. J
Mol Cell Cardiol. 1995;27:383396.
6. Bing OHL, Ngo HQ, Humphries DE, Robinson KG, Lucey EC, Carver
W, Brooks WW, Conrad CH, Hayes JA, Goldstein RH. Localization of
a(I) collagen mRNA in myocardium from the spontaneously hypertensive rat during the transition from compensated hypertrophy to failure.
J Mol Cell Cardiol. 1997;29:23352344.
7. Conrad CH, Brooks WW, Hayes JA, Sen S, Robinson KG, Bing OHL.
Myocardial fibrosis and stiffness with hypertrophy and heart failure in the
spontaneously hypertensive rat. Circulation. 1995;91:161170.
8. Boluyt MO, ONeill L, Meredith AL, Bing OHL, Brooks WW, Conrad
CH, Crow MT, Lakatta EG. Alterations in cardiac gene expression during
the transition from stable hypertrophy to heart failure. Circ Res. 1994;
75:2332.
9. Sirokman G, Humphries DE, Bing OHL. Endogenous retroviral transcripts in myocytes from spontaneously hypertensive rats. Hypertension.
1997;30:88 93.
10. Singh K, Balligand J-L, Fischer TF, Smith TW, Kelly RA. Glucocorticoids increase osteopontin expression in cardiac myocytes and microvascular endothelial cells: role in regulation of inducible nitric oxide
synthase. J Biol Chem. 1995;270:2847128478.
11. Denhardt DT, Guo X. Osteopontin: a protein with diverse functions.
FASEB J. 1993;7:14751482.

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

670

Myocardial Osteopontin

12. Giachelli CM, Schwartz, SM, Liaw L. Molecular and cellular biology of
osteopontin: potential role in cardiovascular disease. Trends Cardiovasc
Med. 1995;5:88 95.
13. Murry CE, Giachelli CM, Schwartz SM. Macrophages express
osteopontin during repair of myocardial necrosis. Am J Pathol. 1994;145:
1450 1462.
14. Ashizawa N, Graf K, Do YS, Nunohiro T, Giachelli CM, Meehan WP,
Tuan T-L, Hsueh WA. Osteopontin is produced by rat cardiac fibroblasts
and mediates AII-induced DNA synthesis and collagen gel contraction.
J Clin Invest. 1996;98:2218 2227.
15. Graf K, Do YS, Ashizawa N, Woerner MP, Giachelli CM, Marbe CC,
Fleck E, Hsueh WA. Myocardial osteopontin is associated with left
ventricular hypertrophy. Circulation. 1997;96:30633071.
16. Fanburg BL, Posner BI. Ribonucleic acid synthesis in experimental
cardiac hypertrophy in rats. Circ Res. 1968;23:123135.
17. Pfeffer JM, Pfeffer MA, Mirsky I, Braunwald E. Regression of left
ventricular hypertrophy and prevention of left ventricular dysfunction by
captopril in spontaneously hypertensive rat. Proc Natl Acad Sci U S A.
1982;79:3310 3314.
18. Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid
guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem.
1987;162:156 159.
19. Singh K, Mukherjee AB, DeVouge M, Mukherjee BB. Differential processing of osteopontin transcripts in rat kidney- and osteoblast-derived
cell lines. J Biol Chem. 1992;267:2384723851.
20. Chen J, Singh K, Mukherjee BB, Sodek J. Developmental expression of
osteopontin mRNA in rat tissues: evidence for a role for osteopontin in
bone formation and resorption. Matrix. 1993;13:113123.
21. Oldberg A, Franzen A, Heinegard D. Cloning and sequence analysis of rat
bone sialoprotein (osteopontin) cDNA reveals an Arg-Gly-Asp cellbinding sequence. Proc Natl Acad Sci U S A. 1986;83:8819 8823.
22. Butler WT. The nature and significance of osteopontin. Connect Tissue
Res. 1989;23:123136.
23. Williams EB, Halpert I, Wickline S, Davidson G, Parks WC, Rottman JN.
Osteopontin expression is increased in the heritable cardiomyopathy of
Syrian hamsters. Circulation. 1995;92:705709.
24. Panda D, Kundu GC, Lee BI, Peri A, Fohl D, Chackalaparampil I,
Mukherjee BB, Li XD, Mukherjee DC, Seides S, Rosenberg J, Stark K,

25.

26.

27.

28.

29.

30.

31.

32.

33.
34.

Mukherjee AB. Potential role of osteopontin and avb3 integrin in the


development of coronary artery restenosis after angioplasty. Proc Natl
Acad Sci U S A. 1997;94:9308 9313.
Kabour A, Henegar JR, Davineni VR, Janicki JS. Prevention of angiotensin II induced myocyte necrosis and coronary vascular damage by
lisinopril and losartan in the rat. Cardiovasc Res. 1995;29:543548.
Giachelli CM, Pichler R, Lombardi D, Denhardt DT, Alpers CE,
Schwartz SM, Johnson RJ. Osteopontin expression in angiotensin
II-induced tubulointerstitial nephritis. Kidney Int. 1994;45:515524.
Calderone A, Thaik CM, Takahashi N, Chang DLF, Colucci WS. Nitric
oxide, atrial natriuretic peptide and cGMP inhibit the growth-promoting
effects of norepinephrine in cardiac myocytes and fibroblasts. J Clin
Invest. 1998;101:812 818.
Patarca R, Freeman GJ, Singh RP, Wei F, Durfee T, Blattner F, Regnier
DC, Kozak CA, Mock BA, Morse HCI, Jerrells TR, Cantor H. Structural
and functional studies of the early T lymphocyte activation 1 (Eta 1) gene:
definition of a novel T cell dependent response associated with genetic
resistance to bacterial infection. J Exp Med. 1989;170:145161.
Balligand J-L, Ungrureanu D, Kelly RA, Kobzik L, Pimental D, Michel
T, Smith TW. Abnormal contractile function due to induction of nitric
oxide synthesis in rat cardiac myocytes follows exposure to activated
macrophage-conditioned medium. J Clin Invest. 1993;91:2314 2319.
Pinskey DJ, Cai B, Yang X, Rodriguez C, Scicca RR, Cannon PJ. The
lethal effects of cytokine induced nitric oxide on cardiac myocytes are
blocked by NOS antagonism or TGF-b. J Clin Invest. 1995;95:667 685.
Haywood GA, Tsao PS, Von der Leyen HE, Mann MJ, Keeling PJ,
Trindade PT, Lewis NP, Byrne CD, Rickenbacher PR, Bishopric NH,
Cooke JP, McKenna WJ, Fowler MB. Expression of inducible nitric
oxide synthase in human heart failure. Circulation. 1996;93:10871094.
Hare JM, Givertz MM, Creager MA, Colucci WS. Increased sensitivity to
nitric oxide synthase inhibition in patients with heart failure: potentiation
of b-adrenergic inotropic responsiveness. Circulation. 1998;97:161166.
Weber KT. Extracellular matrix remodeling in heart failure: a role for de
novo angiotensin II generation. Circulation. 1997;96:4065 4082.
Carver W, Molano I, Reaves TA, Borg TK, Terracio L. Role of the a1b1
integrin complex in collagen gel contraction in vitro by fibroblasts. J Cell
Physiol. 1995;165:425 437.

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

Myocardial Osteopontin Expression Coincides With the Development of Heart Failure


Krishna Singh, Geza Sirokman, Catherine Communal, Kathleen G. Robinson, Chester H.
Conrad, Wesley W. Brooks, Oscar H. L. Bing and Wilson S. Colucci
Hypertension. 1999;33:663-670
doi: 10.1161/01.HYP.33.2.663
Hypertension is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 1999 American Heart Association, Inc. All rights reserved.
Print ISSN: 0194-911X. Online ISSN: 1524-4563

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://hyper.ahajournals.org/content/33/2/663

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Hypertension can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Hypertension is online at:
http://hyper.ahajournals.org//subscriptions/

Downloaded from http://hyper.ahajournals.org/ by guest on August 24, 2015

Anda mungkin juga menyukai